Sie sind auf Seite 1von 47

2 May 2014

EMA/618604/2008 Rev. 9
Committee for Human Medicinal Products (CHMP)

Questions & Answers: Positions on specific questions
addressed to the pharmacokinetics working party
Background
In the context of assessment procedures, the Pharmacokinetics Working Party (PKWP), or its
predecessor the Therapeutic Subgroup on Pharmacokinetics of the Efficacy Working Party (EWP-PK
subgroup), is occasionally consulted by the CHMP or, following CHMPs agreement, by other
Committees, Working parties or the CMD(h). The objective is to address specific questions in relation
to pharmacokinetic evaluations and particularly the requirements and assessment of bioequivalence
studies. The positions, which are being elaborated by the PKWP in response to such questions, are
being forwarded to the enquiring party for consideration in their assessment.
It is understood that such position will be reflected in the procedure-related assessment reports if
applicable. In some cases however, these position might also be of more general interest as they
interpret a very specific aspect that would not necessarily be covered by guidelines. This paper
summarises these positions which have been identified as being within this scope. In addition, general
clarifications related to guidelines authored by the PKWP are subject to specific positions in this paper.
It should be noted that these positions are based on the current scientific knowledge as well as
regulatory precedents. They should be read in conjunction with the applicable guidelines on
bioequivalence in their current version. If the questions have initially been raised in the context of
specific assessment procedures, details of these procedures have been redacted for reasons of
confidentiality.
This compilation will be updated with new positions as soon as they become available. Likewise, if a
position is being considered outdated, e.g. due to new evolutions in the scientific knowledge including
revisions to the applicable guidelines, positions will be removed from this document. Positions
previously prepared by the EWP-PK subgroup are endorsed by the current PKWP unless removed from
this document.
The positions in this document are addressing very specific aspects. They should not be quoted as
product-specific advice on a particular matter as this may require reflection of specific data available
for this product. By no means should these positions be understood as being legally enforceable.
Last update: May 2014

7 Westferry Circus Canary Wharf London E14 4HB United Kingdom
An agency of the European Union

Telephone +44 (0)20 7418 8400 Facsimile +44 (0)20 7418 8613
E-mail info@ema.europa.eu Website www.ema.europa.eu

European Medicines Agency, 2014. Reproduction is authorised provided the source is acknowledged.

Table of contents

1. Requirements for food-interaction studies for modified release
formulations .......................................................................................... 3
2. Bioequivalence studies in children ........................................................ 7
3. Bioequivalence of gastro-resistant preparations (e.g.
omeprazole) .......................................................................................... 8
4. Bioequivalence studies for generic products containing
clopidogrel .......................................................................................... 10
5. Acceptance criteria for bioequivalence studies for losartan ................ 13
6. Bioequivalence assessment of generics for tacrolimus ........................ 14
7. Requirements for demonstration of bioequivalence for
ciclosporine generics ........................................................................... 16
8. Requirements for demonstration of bioequivalence for
mycophenolate mofetil generics .......................................................... 17
9. Recommendations on determination of absolute and relative
bioavailability ...................................................................................... 19
10. Clarification on the recommended statistical method for the
analysis of a bioequivalence study ...................................................... 20
11. Requirements for demonstration of bioequivalence for generics of
biphasic modified release formulations for oral use ............................ 32
12. Effect of sorbitol on the pharmacokinetics of highly permeable
drug substances .................................................................................. 33
13. Requirement to perform incurred sample reanalysis ........................... 35
14. Number of subjects in a two-stage bioequivalence study design ........ 38
15. Bioequivalence studies for generic application of omega 3 fatty
acid ethylesters in a soft gelatine capsule .......................................... 39
16. Acceptability of an additional strengths biowaiver when
bioequivalence to the reference product has been established
with a BCS-based biowaiver ............................................................... 41
17. Question on a generic application for Quetiapine Lambda 200,
300, 400 mg prolonged release tablets .............................................. 42
18. BCS classification of memantine ......................................................... 45
19. Ebastine: use of metabolite data to demonstrate bioequivalence
between inactive pro-drugs NEW ........................................................ 46


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 2/47

1. Requirements for food-interaction studies for modified release
formulations
The position of the EWP PK Group is as follows:
a. Guideline recommendations (CPMP/EWP/280/96) and general aspects
Food interactions may be related to the drug substance itself and/or the formulation, the latter being
most important in the case of modified release (MR) products.
The aim of food effect studies for new MR formulations (developed either for a new substance or for a
substance previously approved in an instant release formulation) is to evaluate the influence of food on
the absorption of the drug substance from the new formulation, to evaluate the clinical relevance of a
potential food effect and when needed to provide appropriate dose recommendations with respect to
intake of the product in relation to meals. This is clearly stated in paragraph 4.1.4.1 of the guideline:
Different modified release formulations of the same drug substances may differ with respect
to food interaction. Hence, the influence of food on the bioavailability of oral modified release
formulations must be investigated for safety and efficacy purposes.
The optimal experimental conditions to produce a food effect include the ingestion of a
predefined high fat meal immediately before dosing. For the assessment of food effects besides
AUC and Cmax, it may also valuable to compare the modified release characteristics.
If a significant food effect is found, applicant should give a justified dose recommendation with
respect to the intake of the product in relation to meals.
Possible approaches for the investigation of the effect of food on the bioavailability of modified
release forms reflecting the present scientific approach are presented in Annex 1. However,
due to the complexity of the food-drug interaction with any particular dosage form a different
approach for in vivo studies can be accepted if adequately justified.
Food effect studies for new MR formulations should be conducted early during drug development so
that appropriate recommendations regarding intake in relation to food can be included in clinical
efficacy and safety studies.
In contrast to new MR formulations, for generic MR products bioequivalence under fed conditions is
required rather than the investigation of food interaction as described in paragraph 4.1.4.1, i.e.
- paragraph 5.1 reg. prolonged release formulations states that the effect of food on the in vivo
performance is comparable for both formulations when a single dose study is conducted
comparing equal doses of the test formulation with those of the reference formulations
administered immediately after a predefined high fat meal. This study should be conducted
with the same strength as those of the pivotal bioequivalence studies.
- paragraph 5.2 regarding delayed release formulations states that As food can influence the
absorption of an active substance administered in an enteric-coated formulation, post-prandial
bioequivalence studies are necessary.
It has been shown that food composition (fat content) and timing may be crucial for drug product
bioavailability. Administration immediately after completing a high fat meal serves as kind of worst
case situation in terms of product performance/robustness. Therefore, a food interaction study should
be performed accordingly.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 3/47

Section 4.1.5.1 of the guideline states
If the modified release formulation contains a higher dose compared to the approved
immediate release product, the possibility of unexpected release resulting in unacceptable
higher exposure should be excluded.
One issue that is important to consider for both new MR formulations and generic MR formulations is
the influence of alcohol on the MR formulation and the risk for unexpected release caused by alcohol
ingestion.
b. Study design - Guideline recommendations (CPMP/EWP/280/96) based on App. 1
Appendix 1 of the guideline provides recommendations regarding study design in different scenarios.
Some explanation and comments to these recommendations are given below.
Bioanalytical measurements should include quantification of metabolites or enantiomers if respective
requirements apply.
1. MR formulation developed for a New Chemical Entity (NCE)
For MR formulations developed for an new chemical entity the guideline recommends a single dose
4 way crossover study ; MR fed and fasted + oral solution (or immediate release (IR) formulation if
a solution is not feasible) fed and fasted. With this study design the effect of food on both the
substance and the MR formulation can be evaluated.
However section 4.1.4.1 of the guideline also states that a different approach for in vivo studies
can be accepted if adequately justified. Hence, a 2-way cross over study (MR formulation fasting
and fed) could be sufficient to evaluate the formulation related food effect.
The guideline also states that a single dose 3 way crossover study may be required in case the
clinical trial formulation differs from the to-be-market product; i.e. comparing clinical trial
formulation fasted with to-be-marketed formulation fed and fasted. However, if there is a marked
food effect on the clinical trial formulation and the formulation has been taken under non-fasting
conditions in the clinical studies, it may be advantageous to have comparative data on the food-
effect on the marketing formulation in the same study, i.e. also here a 4-way crossover study with
clinical trial and marketing formulation under fasting and fed conditions. This information may be
important in the evaluation of dosing recommendations.
In case there is a marked food-effect, additional food-interaction studies might be needed to
support dosing recommendations, i.e. studies of the effect of different kinds of food, studies
investigating the effect of a meal taken at certain time period before and after the drug, etc.
2. MR formulation developed after an approved IR formulation
The guideline recommends a single dose 3 way crossover study; MR fed and fasted + IR fasted.
However, the design of this study depends on which other studies that are conducted comparing
the new MR formulation with the approved IR formulation and if there is a clinically significant food
effect on the IR formulation. If there is no food effect on IR formulation, a 2-way cross-over study
comparing MR formulation fasted and fed could be sufficient (given that other studies compare the
MR and IR formulation under fasting conditions). In case of a clinically significant food effect for
the IR formulation, a 4-way cross-over study comparing MR formulation fasted and fed and IR
formulation fasted and fed could be useful to quantify the food effect on each formulation. If a 3-
way cross-over study is conducted with IR formulation in one arm, consideration should be given
to whether the IR formulation should be taken fasted or in a fed state (i.e. intake in accordance
with the recommendation in the SPC).

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 4/47

3. MR formulations developed as generics
For generic products, the guideline recommends two single dose 2 way crossover studies
evaluating test and reference fasted, and test and reference fed, respectively. Alternatively a single
dose 4 way crossover study (MR generic fed and fasted + reference fed and fasted) can be
conducted to demonstrate bioequivalence between generic and reference in both fasting and fed
state. In a 4 way crossover study a comparison of the food effect for test and reference is possible,
which will not be the case if two 2 way cross over studies are conducted, as between study
comparison of food effect is not recommended.
For both single-unit formulations and multiple-unit formulations, the highest strength should in general
be studied. In case a non-linearity in the food effect is suspected, the food interaction study should be
performed with the highest and the lowest strength.
c. Defining a high fat meal
Presently, the guideline on modified release formulations does not give any advise regarding the type
of meal, but the composition of a high fat meal meal is recommended in the revised Guideline on the
Investigation of Bioequivalence (CPMP/QWP/EWP/1401/98 Rev. 1) as follows:
the meal should be high fat (approximately 50 percent of total caloric content of the meal) and high-
calorie (approximately 800 to 1000 kcal) meal. This test meal should derive approximately 150, 250,
and 500-600 kcal from protein, carbohydrate, and fat, respectively. The composition of the meal
should be described with regard to protein, carbohydrate and fat content (specified in grams, calories
and relative caloric content (%)).
d. Evaluation
Evaluation of food study results includes metabolites or enantiomers in case respective requirements
apply.
New MR formulations
For MR formulations developed for a NCE or MR products developed after an approved IR formulation
the food interaction study will provide quantitative data on the extent of influence of food on the
pharmacokinetics. The clinical relevance of the effect of food should be discussed both from an efficacy
and a safety perspective. When needed dose recommendations with respect to intake of the product in
relation to meals should be given. Additional studies with other types of food, or with intake of the
drug at certain time intervals before and after a meal may be needed to support the proposed dose
recommendations.
Generic MR formulation
The bioequivalence approach considering usual acceptance limits (80 125 %) is applicable for generic
MR products. If bioequivalence between generic and reference has been demonstrated both in fasting
and in fed state the MR generic product and the reference can be considered to behave similar under
fed conditions.
Any widening of the acceptance criteria for Cmax should follow the recommendations of the revised
guideline on the investigation of bioequivalence (CPMP/EWP/QWP/1401/98 rev 1).
For delayed release formulations with single unit dosage forms differences in tmax is also
recommended to be assessed, especially for products where a fast onset of action is important.


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 5/47

e. Special cases
1. Can a MR product be considered a generic if it has no food-effect as opposed to the innovator
which has one?
In general a generic is meant to be bioequivalent with the innovator under fasted and fed
conditions. A difference regarding formulation related food interactions indicates product
differences thus contradicting the generic by definition. Accordingly, for products where
bioequivalence can be shown in the SPC recommended condition but not in the non-
recommended state due to a different food effect, the product does not fulfil the requirements
of a generic product, but could be eligible for an Article 10(3) application.
2. What studies are needed for a generic if the innovators SPC states that it should be taken with
a meal only or only in the fasted state?
Comparative studies should be performed under both fed and fasted conditions. See also
response above.

Since the guideline on modified release formulations (CPMP/EWP/280/96) is currently under revision
certain requirements may be changed with a revised document.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 6/47

2. Bioequivalence studies in children
The EWP-PK subgroup was asked to address the following questions: Treatment of children often
requires that new formulations or strengths are developed. If chemical-pharmaceutical data are not
considered sufficient to establish bioequivalence should bioequivalence studies be conducted in children
or would healthy volunteers suffice?
The position of the EWP-PK subgroup is as follows:
In vivo bioequivalence is almost always established in healthy volunteers unless the drug carries safety
concerns that make this unethical. This model, in vivo healthy volunteers, is regarded adequate in
most instances to detect significant formulation differences and the results will allow extrapolation to
populations in which the drug is approved (the elderly, patients with renal or liver impairment etc). The
same reasoning applies also to children. Hence, in the vast majority of cases BE studies in healthy
volunteers are adequate for products intended for use in children.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 7/47

3. Bioequivalence of gastro-resistant preparations (e.g. omeprazole)
The EWP-PK subgroup was asked to address the following question: What are the recommendations
for demonstration of bioequivalence of gastro-resistant preparations (e.g. omeprazole)?
The position of the EWP-PK subgroup is as follows:
General aspects:
According to section 5.2 Delayed release formulations of the Note for Guidance on Modified Release
Oral and Transdermal Dosage Forms (CPMP/EWP/280/96), in gastro-resistant or enteric products
bioequivalence should be demonstrated not only in a single dose study in fasted conditions, but also in
a single dose study under fed conditions. The fed study should be conducted using a high-fat meal
(approximately 50 percent of total caloric content of the meal) and high-calorie (approximately 800 to
1000 kcal) meal. This test meal should derive approximately 150, 250, and 500-600 kcal from protein,
carbohydrate, and fat, respectively. The composition of the meal should be described with regard to
protein, carbohydrate and fat content (specified in grams, calories and relative caloric content (%)).
Consequently, bioequivalence studies should be performed under both fed and fasting conditions. In
general a generic is meant to be bioequivalent with the innovator under fasted and fed conditions. A
difference regarding formulation related food interactions indicates product differences thus
contradicting the generic by definition. Accordingly, for products where bioequivalence can be shown in
the SPC recommended condition but not in the non-recommended state due to a different food effect,
the product does not fulfil the requirements of a generic product, but could be eligible for an Article
10(3) application. See also section 2 Requirements for food-interaction studies for modified release
formulations for recommendations regarding study design, etc.
Gastric emptying of single unit dosage forms non-disintegrating in the stomach (e.g. enteric coated
tablets) is prolonged and highly erratic, most likely due to the effect of the inter-digestive cycle within
the Migrating Myoelectric Complex. The consequences of this effect on the enteric coating of delayed
release formulations are largely unpredictable: if e.g. the active pharmaceutical ingredient (API)
release occurs prior to stomach emptying because of prolonged residence in the stomach either
degradation can occur or the release may be considerably delayed. In either case erratic concentration
profiles (either non-existing or extremely delayed) can be obtained. Therefore the sampling period
should be designed such that measurable concentrations are obtained, taking into consideration not
only the half-life of the drug but the possible occurrence of this effect as well. This should reduce the
risk of obtaining incomplete concentration-time profiles due to delay to the most possible extent. These
effects are highly dependent on individual behaviour. Therefore, but only under the conditions that
sampling times are designed to identify very delayed absorption and that the incidence of this outlier
behaviour is observed with a comparable frequency in both, test and reference products, these
incomplete profiles can be excluded from statistical analysis provided that it has been considered in the
study protocol.
The general requirements for biowaiver of an additional strength detailed in section 4.1.6 of the
Guideline on the investigation of bioequivalence (CPMP/EWP/QWP/1401/98 rev 1) are applicable also
for delayed release tablets and recommendations regarding which strength to study is given in the
same section of this guideline and also in section 2 Requirements for food-interaction studies for
modified release formulations. When evaluating proportionality in composition, it is recommended to
consider the proportionality of gastro-resistant coating with respect to the surface area (not to core
weight) to have the same gastro-resistance (mg/cm
2
).

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 8/47

The dissolution profiles should be compared not only in Pharmacopoeial conditions (2 hours at pH 1.2
followed by 45 minutes at pH 6.8), but also at more neutral pHs in the range 2-5, both for single unit
non disintegrating and disintegrating dosage forms with multiple units. Hence, at least, two dissolution
tests in two steps are required. First a comparison at pharmacopoeial conditions, 2 hours at pH 1.2
followed by 45 minutes in pH 6.8 and then a second separate dissolution test at a higher initial pH
mimicking fed state e.g. 2 hours at 4.5 followed by 45 minutes in pH 6.8.
Concluding similarity if dissolution of more than 85% is obtained within 15 minutes is not applicable for
gastro-resistant formulations. In case of gastro-resistant formulations the release occurs after gastric
emptying (median approx. 13 15 min). Therefore, the comparison of dissolution profiles should be
performed even if dissolution is more than 85% before 15 min in either products or strengths. Hence,
a tight sampling schedule is recommended after the product has been investigated for 2 h in media
mimicking the gastric environment (pH 1.2 or 4.5) since profile comparison (e.g. using the f2
calculation) is required.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 9/47

4. Bioequivalence studies for generic products containing clopidogrel
The platelet aggregation inhibitor clopidogrel is pre-systemically hydrolysed to the inactive metabolite
clopidogrel carboxylic acid. The plasma levels of the unchanged drug are up to 2000 fold lower than
those of the carboxylic acid metabolite. Another metabolite, clopidogrel thiol, formed by a parallel
pathway, is the pharmacologically active form of clopidogrel and is generated in the intestine and liver
primarily by the CYP2C19 enzyme isoform. Due to its chemical instability and low circulating levels, its
detection in plasma is problematic. Clopidogrel thiol irreversibly binds to the P2Y12 receptors of ADP
on the platelet membranes in portal and systemic circulation, leading to the inhibition of platelet
aggregation.
During the evaluation of the Marketing Authorisation applications for generic product of clopidogrel, the
following questions were addressed by the CHMP to the EWP-PK subgroup and the EWP-CVS subgroup
group
1
, respectively:
1. Which substance should be studied in bioequivalence studies: the parent compound clopidogrel
or the metabolite(s) of clopidogrel?
The Guideline on the investigation of bioequivalence (CPMP/EWP/QWP/1401/98 Rev 1) states Also for
inactive prodrugs, demonstration of bioequivalence for parent compound is recommended. The active
metabolite does not need to be measured.
At the time of approval of the reference product Plavix, no reliable and validated methodology for the
determination of the pharmacokinetics of the parent prodrug clopidogrel or of the active metabolite
clopidogrel thiol was available. Thus, at the time, the pharmacokinetic profile of clopidogrel was
established based on the pharmacokinetics of clopidogrel carboxylic acid, which is the non-active
metabolite. In the meantime, the pharmacokinetic profile characterisation of clopidogrel has improved
by development of a sensitive analytical technique (e.g. LC-MS-MS) enabling for a suitable
investigation of the parent prodrug, clopidogrel. A more accurate picture of the PK profile of clopidogrel
can be obtained.
Position of the EWP-PK subgroup:
The demonstration of bioequivalence between the reference and the generic compound should be
based on the parent prodrug, clopidogrel.
2. Is demonstration of bioequivalence under fed conditions necessary in addition to the
demonstration under fasting conditions?
At the time the innovative drug-product was developed, no data regarding the effect of food on the
bioavailability of clopidogrel parent compound were available. More recently, the investigation of food
intake influence on the bioavailability of clopidogrel has been investigated. The results obtained by
Nirogi et al. (Nirogi, RV et al., Arzneimittelforschung 2006; 56(11); 735-9: Effect of food on
bioavailability of a single oral dose of clopidogrel in healthy male subjects) indicate that in the fed state
the bioavailability of a single oral dose of clopidogrel increases dramatically (500 - 600 %) but the
systemic exposure to the major but inactive carboxylic acid metabolite increases only by approximately
10-20 %. The current Summary of Product Characteristics (SPC) for the originator states that
clopidogrel should be given as a single daily dose of 75 mg with or without food.
1
EWP Therapeutic Subgroup on Cardiovascular Issues

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 10/47


Position of the EWP-PK subgroup:
The Guideline on the investigation of bioequivalence (CPMP/EWP/QWP/1401/98 Rev 1) states In
general, a bioequivalence study should be conducted under fasting conditions as this is considered to
be the most sensitive condition to detect a potential difference between formulations. For products
where the SmPC recommends intake of the reference medicinal product on an empty stomach or
irrespective of food intake, the bioequivalence study should hence be conducted under fasting
conditions.
The food effect on the bioavailability (BA) of the unchanged clopidogrel - not recognised in the SPC -
was not investigated by the innovator before approval of the originator product since a sensitive
analytical method was not available at the time of approval. However, a publication by Nirogi et al.
(2006) suggested a significant food effect with a high-fat meal. Similar results have been observed in
applications for generic medicinal products. The food effect might be due to a protection from acidic
hydrolysis in the stomach in a fasting state, since the BA is enhanced under fed conditions. The EWP-
PK subgroup reviewed the solubility properties of clopidogrel salts and these indicate that when
administration of clopidogrel occurs under fasting conditions, the dissolution in the gastric media with a
subsequent hydrolysis and formation of the inactive carboxy-acid metabolite is maximal. As a
consequence, the extent of unchanged drug that still is available for absorption (at the intestine level)
is reduced. Conversely, the dissolution of clopidogrel is limited in the gastric media under fed
conditions, the acidic hydrolysis in the stomach is reduced and the BA of clopidogrel is improved.
The EWP-PK subgroup acknowledges that as a consequence, the solubility of salts might be important.
However, all clopidogrel salts have high solubility at low pH and the risk for acidic hydrolysis may
therefore be similar. The food effect could consequently be expected to be similar to the reference
product for different salts. Hence, the EWP-PK subgroup considered that there was currently an
insufficient scientific rationale to justify a deviation from the revised bioequivalence guideline and
bioequivalence should be demonstrated under fasting conditions irrespective of the salt.
Should further information on the food effect of clopidogrel become available, the SPC would be
amended accordingly.
3. Bioanalytical methods: Should there be any special requirements to ensure that the risk of
back-conversion of the major metabolite to clopidogrel could be excluded?
Within several centralised clopidogrel applications, the CHMP raised concerns about the possible back-
conversion of the major metabolite of clopidogrel (clopidogrel carboxylic acid) to clopidogrel during the
bio-analytical analysis of the samples. Considering that plasma levels of clopidogrel carboxylic acid
observed in patients or healthy volunteers treated with clopidogrel are much higher than that of the
parent drug, a minimum back-conversion of the metabolite could potentially lead to a huge over-
estimation of clopidogrel plasma levels and would bias the outcome of bioequivalence study.
Position of the EWP-PK subgroup:
The EWP-PK subgroup confirmed that back-conversion could potentially occur when methanol is used
as (part of) extraction solvent, reconstitution solvent, chromatography mobile phase or for the
preparation of calibrators, quality control (QC) solutions and internal standards during bioanalysis.
Therefore, testing for the back-conversion of clopidogrel carboxylic acid metabolite should be part of
the validation process of analytical methods used for the measurement of clopidogrel plasma levels.
It should be demonstrated that there is no back-conversion of the major metabolite to the parent drug
clopidogrel under all conditions for sample handling (including extraction procedures) and storage.


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 11/47

4. Could the acceptance criteria for C
max
be widened?
According to the Guideline on the investigation of bioequivalence (CPMP/EWP/QWP/1401/98 Rev 1)
widening of the acceptance criteria for C
max
is possible for highly variable drug products provided that
a wider difference in C
max
is considered clinically irrelevant based on a sound clinical justification. The
revised bioequivalence guideline provides detailed advice on how the acceptance criteria can be
widenened for highly variable drug products with a bioequivalence study of replicate design and using
the scaled-average-bioequivalence approach. However, a prerequisite for widening the acceptance
criteria is that a wider difference in C
max
is considered clinically irrelevant. This issue was assessed by
the EWP-CVS subgroup.
Position of the EWP-CVS subgroup:
The EWP-CVS subgroup evaluated the request from widening the 90% confidence interval for C
max

from the efficacy and safety perspectives. The EWP-CVS subgroup considered what would be the
degree of the impact of the possible variations in the C
max
following the 75 mg dose, since some data
suggest the existence of a plateau response in the inhibition of platelets aggregation. However, it is
currently not entirely clear what would be the influence of variable clopidogrel concentrations on
pharmacodynamics. It is important to note that clopidogrel is approved and recommended for use in
acute clinical conditions, for which a high loading dose is advised in order to attain a fast antiplatelet
action. Whether in these situations a lower C
max
might be of clinical relevance is unknown, but cannot
be completely excluded.
In conclusion, it is not definitely proven that widening C
max
acceptance range for clopidogrel is devoid
of clinically relevant implications, both in terms of safety and efficacy, for all situations where the drug
is used in clinical practice. Under these circumstances, the widening of 90% confidence intervals for
C
max
is not recommended.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 12/47

5. Acceptance criteria for bioequivalence studies for losartan
The EWP-PK subgroup was asked to address the following question: Which analyte, parent and/or
metabolite, should be used for the decision of bioequivalence in the case of losartan, and which
acceptance criteria should be applied.
Position of the EWP-PK subgroup:
Losartan is not a pro-drug. It is an angiotensin II antagonist at the AT1-subtype receptor. In humans,
losartan competitively binds to the AT1 receptor, while the metabolite E3174 binds non-competitively.
The active metabolite E3174 is not directly formed from losartan, but from an intermediate product,
metabolite E3179. Alternatively, the E3179 intermediate can also be hydroxylated to an inactive
metabolite. It has been estimated that about 14% of the orally administered losartan dose is converted
into E3174. In addition, 5 other minor metabolites exists that exhibit activity but much less than
parent.
AUC of the active metabolite is 4 8 fold higher than parent, as it is cleared about 10-fold slower than
parent.
Plasma free fractions of parent are 1.3% and that of the active metabolite 0.2%. Losartan and its
metabolite E3174 shows linear pharmacokinetics.
It has been shown in vitro that the IC50 for binding to the AII receptor in smooth muscle cells is 10-
fold more potent for the metabolite than parent and that the in vitro AII concentration dependent
contractile response in rabbit aorta is 33-fold higher for the metabolite. In vivo, in normotensive and
renal hypertensive rats, the active metabolite has been shown to be 15 20-fold more potent
compared to the parent.
Based on in vivo studies in rat, in which the potency was 15 20-fold higher for the metabolite, and
assuming a more or less comparable protein binding as that observed for human plasma (literature
indicated for losartan a binding >99% in rat plasma), the metabolite activity is about 76 100-fold
higher than the parent compound.
Hence, based on total exposure (AUC), the metabolite accounts for the majority of the activity.
However, losartan and the active metabolite have different plasma-concentration time course, with
considerably higher losartan plasma concentrations during the first hours after administration.
Considering the plasma concentration time course, difference in activity and protein binding, losartan
may account for a large part of the activity during the first hour after the first drug administration, and
at losartan tmax, which occur after about one hour, contribution to activity may be almost equal for
losartan and the metabolite. Thereafter, the metabolites contribution to activity is much larger.
Moreover, as the active metabolite E3174 is formed via an intermediate product and not direct from
the parent, the pharmacokinetic data for metabolite E3174 may not reflect the rate of absorption of
parent.
Therefore, bioequivalence for losartan should be proven based upon parent data. Regarding what
acceptance criteria to apply, the submitted documents do not allow any conclusion to be drawn on this
and consequently a conservative approach using 90% CI of 80 125% for AUC and Cmax applies.


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 13/47

6. Bioequivalence assessment of generics for tacrolimus
In relation to the bioequivalence guideline, which has been drafted by the EWP-PK subgroup, a
question was raised regarding the assessment of bioequivalence for tacrolimus generic products. There
were different views whether the normal (80-125%) or a tightened (90-111%) acceptance range for
the 90% CIs, for both AUC and Cmax, should be applied.
The decision on the bioequivalence criteria requires the clinical judgement whether
tacrolimus is considered a narrow therapeutic index drug (NTID). Therefore, the response to
this question has been prepared by the Efficacy Working Party (EWP) taking the EWP-PKs
general position on bioequivalence criteria for NTIDs into account.
The position of the EWP is as follows:
The decision on whether a particular active substance may be considered to be a narrow therapeutic
index drug (NTID), and whether narrowing of the bioequivalence acceptance limits should apply, needs
to be based on clinical considerations of the dose- or concentration-response relationships for both
efficacy and safety.
The following key issues are identified for tacrolimus:
Tacrolimus is a drug that requires individual dose titration to achieve a satisfactory balance
between maximizing efficacy and minimizing serious dose related toxicity. Plasma level
monitoring is routinely employed to facilitate dose titration.
Recommended Therapeutic Drug Monitoring schemes often set desirable levels close to the
upper or lower limit of the therapeutic window (5 ng/ml or 20 ng/ml), for example the use of
minimisation protocols using low levels during maintenance phase. It is well established from
clinical experience with the drug that even small changes of dose can lead to crossing the
upper or lower limits of the therapeutic window
In the case of kidney and heart transplantation, there is only a two fold difference in the upper
and lower limit of the proposed therapeutic range (whole blood levels from 10 to 20 ng/mL).
This is comparable to the therapeutic range for classical NTIDs such as digoxin.
The consequences of over-dosing and of under-dosing (including morbidity/mortality
associated with graft rejection) are of major clinical importance and can substantially affect
clinical outcome.
For the above reasons the EWP considers that tacrolimus is a drug with a narrow therapeutic index.
In a number of EU countries generic prescribing is the norm and pharmacies may dispense either the
branded product or a generic. Where multiple generics are available patients may be switched from
one generic to another when renewing their prescription. Changes of formulation in this situation would
not normally be accompanied by re-titration. The usual frequency of whole blood drug level
measurements in clinical practice (typically once per month during maintenance phase) is not
sufficiently frequent to ensure avoidance of over or under dosing as a result of a patient switching to a
different formulation in the event of generic substitution of tacrolimus. Therefore, in order to ensure
the safety and efficacy of generic tacrolimus products it is necessary to apply tighter bioequivalence
acceptance criteria than the conventional 80-125%.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 14/47

The EWP discussion also covered whether the narrowing of the bioequivalence acceptance criteria to
[90-111%] can be limited to AUC and will not be needed for C
max
. For tacrolimus, this is supported by
the following PK and PK/PD characteristics. Total drug exposure (AUC) is considered to be the key
parameter of importance for dose titration of tacrolimus; in comparison peak whole blood levels do not
seem to be critical for either safety or efficacy. As tacrolimus has a long elimination half life C
min

trough levels can be used as a surrogate for AUC in clinical practice. Given the long terminal half-life,
tacrolimus accumulates during repeated dosing. Due to this accumulation, a potential difference
between formulations in C
max
after single dosing can be expected to be less at steady state, if AUC is
the same for the two formulations. Therefore, the normal acceptance criteria for C
max
[80-125%] can
be used in single dose bioequivalence studies for tacrolimus.
Conclusion: The EWP recommends that the bioequivalence acceptance criteria for tacrolimus should be
[90-111%] for AUC and [80-125%] for C
max
.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 15/47

7. Requirements for demonstration of bioequivalence for ciclosporine
generics
The reference product Neoral soft gelatine capsule concerns a specific formulation of ciclosporin which
undergoes microemulsification process at administration (in the presence of water). For Neoral, the
SmPC indicates a 26% decrease in Cmax and a 15% decrease in AUC, in case the product is taken with
a high fat meal.
As indicated in the guideline on bioequivalence (CPMP/EWP/QWP/1401/98 Rev 1.), for products with
specific formulation characteristics, like Neoral, bioequivalence studies performed under both fasted
and fed conditions are required unless the product must be taken only in the fasted state or only in the
fed state. Neoral may be taken with or without food, and in clinical practice, ciclosporin is often
recommended to be taken in a standardised way in relation to food. Hence, a generic ciclosporin
product must be bioequivalent with the originator product both in fasting and in fed state.
As EWP has defined ciclosporine to be a NTID, for which both AUC and Cmax are important for safety
and efficacy, a narrowed (90.00-111.11%) acceptance range should be applied for both AUC and
Cmax, under fasting as well as under fed conditions, in line with the guideline on bioequivalence
(CPMP/EWP/QWP/1401/98 Rev 1.).
Although a generic product with a reduced food effect could be considered an improvement, this would
not be considered acceptable for a generic application, but could be considered for a hybrid
application, article 10(3) with additional data to support an application under this legal basis.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 16/47

8. Requirements for demonstration of bioequivalence for
mycophenolate mofetil generics
The CMDh requested from the PKWP a position concerning interpretation of the revised Guideline on
the Investigation of Bioequivalence with respect to the bioequivalence data for inactive pro-drugs in
relation to both parent drug and metabolite in the context of demonstration of bioequivalence for
mycophenolate mofetil.
The questions relate to the circumstances under which it is acceptable to base bioequivalence
decision solely on metabolite data if a pro-drug plasma level is measurable. The revised guideline
states: Also for inactive pro-drugs, demonstration of bioequivalence for parent compound is
recommended.
1) If the exact meaning of the word recommended in the context of mycophenolate mofetil (MMF),
depends on
either the feasibility of the technical detection limits, in which the concentrations of the
inactive prodrug are approximately 12000- to 6000-fold lower, for AUC and C
max
,
respectively, than that of the active metabolite mycophenolic acid.
or should specific PK-parameters be taken into account, low exposure of the parent
resulting in a short Tmax, which makes it not relevant to measure the parent drug.
Position of the PKWP:
The bioequivalence guideline states for inactive prodrugs, demonstration of bioequivalence for parent
compound is recommended. The guideline further clarifies: However, some pro-drugs may have low
plasma concentrations and be quickly eliminated resulting in difficulties in demonstrating
bioequivalence for parent compound. In this situation it is acceptable to demonstrate bioequivalence
for the main active metabolite without measurement of parent compound. Hence, although the
guideline recommends the use of parent compound also for inactive pro-drugs, exceptions are
possible. The acceptability of use of main active metabolite instead of parent compound will be
determined based both on the feasibility of measuring parent compound and on the pharmacokinetic
characteristics for parent compound and active metabolite. For pro-drugs with a very large difference
in exposure between parent and active metabolite and where the pro-drug is quickly eliminated, it is
expected that there can be difficulties in demonstrating bioequivalence for parent compound and
demonstration of bioequivalence based on active metabolite alone can be accepted.
For mycophenolate mofetil (MPM) specifically, the parent compound undergoes extensive presystemic
metabolism to the active metabolite MPA. Moreover, MPM half-life is very short (0.60 to 1.20 h as
reported) resulting in approximately 12000- and 6000-fold lower AUC and C
max
respectively, for parent
compound compared to metabolite. MPM has a tmax of 0.5 h and a t1/2 of less than 1 h, which limits
the characterisation of the early plasma concentrations. As a consequence reliable estimation of Cmax
will be difficult. In this situation it is acceptable to demonstrate bioequivalence for the main active
metabolite without measurement of parent compound as stated in the bioequivalence guideline.


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 17/47

2) Is it acceptable NOT to follow this recommendation and use ONLY metabolite data to demonstrate
bioequivalence between two products of the same pro-drug mycophenolate mofetil, even when
current analytical assays allow measuring the parent with acceptable sensitivity?
Position of the PKWP:
A recommendation leaves room for an exceptional decision on a case by case basis. In this case it is
clear that the parent compound is inactive and completely converted into the active metabolite yielding
a 12000 fold difference in AUC. Due to this, demonstration of bioequivalence between two products of
the same pro-drug can be based on metabolite data only. The argument that current analytical assays
allow measuring the parent with acceptable sensitivity cannot be readily taken considering the short
tmax and t1/2 of the parent compound which will limit a reliable estimation of Cmax of the parent
compound.


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 18/47

9. Recommendations on determination of absolute and relative
bioavailability
Absolute bioavailability
Information on absolute bioavailability is important in the overall evaluation of the pharmacokinetics of
the drug substance. For some new chemical entities information on absolute bioavailability facilitates
the evaluation of the mass balance study, and enables conclusions regarding the contribution of
different elimination routes to drug clearance. This information is important when determining the
need for studies in subjects with renal and hepatic impairment as well as the need for drug-drug
interaction studies at biliary excretion level. The information is also useful when predicting the
consequences of pre-systemic drug-drug interactions, both at absorption and metabolism level.
Therefore, for new active substances intended for systemic action, the absolute bioavailability should,
if possible, be determined by comparing the bioavailability of the intended pharmaceutical form for an
extra-vascular route of administration with an intravenous administration. For substances with non-
linear pharmacokinetics, consideration should be given to the dose(s) used for evaluation of absolute
bioavailability. Furthermore, data on absolute bioavailability is valuable in the evaluation of BCS based
biowaivers (see Guideline on the investigation of bioequivalence, CPMP/EWP/QWP/1401/98 Rev. 1).
Relative bioavailability
It is recommended to obtain information on the relative bioavailability of different dosage forms (or
formulations) used during drug development. By definition relative bioavailability is the comparison of
different dosage forms (or different formulations thereof) administered by the same or a different non-
intravenous route (e.g. tablets vs. oral solution).
Regarding formulation changes during drug development, unless BCS based biowaiver is applicable
bioequivalence studies are needed if there has been a change between the formulation used in phase
III and the final marketing formulation which may affect rate or extent of absorption. Relative
bioavailability studies (or comparative bioavailability studies) are recommended between different
formulations used during phase I, II and III. There is no requirement for demonstration of
bioequivalence between phase II and phase III formulations. It is assumed that any difference in rate
or extent of absorption between these formulations is taken into account in the design of the phase III
studies. The clinical relevance of any differences in exposure between formulations used in phase I, II
and III studies should be discussed in applications for NCEs in Module 2.5 and 2.7.1 and taken into
account in the assessment of pharmacokinetic data in Module 2.7.2.

Recommendations for Suprabioavailable products
A suprabioavailable product displays appreciably larger extent of absorption than an approved
reference medicinal product.
If suprabioavailability is found, development of a lower dosage strength should be considered. In this
case, the biopharmaceutical development should be reported and a final comparative bioavailability
study comparing the reformulated new product with the approved reference medicinal product should
be submitted. The potential for a difference in food effect on the rate and/or extent of absorption or a
difference in absorption interactions between the reformulated new product and the approved
reference product should be discussed and when relevant evaluated in vivo.
In case a lower dosage strength has not been developed the dosage recommendations for the
suprabioavailable product will have to be supported by clinical studies.


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 19/47

10. Clarification on the recommended statistical method for the analysis
of a bioequivalence study
1. Introduction
The following text on the general analysis of bioequivalence studies is included in the guidance
document. The bold text is the main sentence of interest for this discussion.

4.1.8 Evaluation
Statistical analysis
The assessment of bioequivalence is based upon 90% confidence intervals for the ratio of the
population geometric means (test/reference) for the parameters under consideration. This method is
equivalent to two one-sided tests with the null hypothesis of bioinequivalence at the 5% significance
level.
The pharmacokinetic parameters under consideration should be analysed using ANOVA. The
data should be transformed prior to analysis using a logarithmic transformation. A confidence interval
for the difference between formulations on the log-transformed scale is obtained from the ANOVA
model. This confidence interval is then back-transformed to obtain the desired confidence interval for
the ratio on the original scale. A non-parametric analysis is not acceptable.
The precise model to be used for the analysis should be pre-specified in the protocol. The statistical
analysis should take into account sources of variation that can be reasonably assumed to have an
effect on the response variable. The terms to be used in the ANOVA model are usually sequence,
subject within sequence, period and formulation. Fixed effects, rather than random effects,
should be used for all terms.

Following the publication of revised version of the Guideline on the Investigation of Bioequivalence
(CPMP/QWP/EWP/1401/98 Rev.1) this paragraph raised several questions from interested parties. The
reason for this interest was twofold. Firstly, the new guideline gives more emphasis to replicate design
trials and evaluation of such trials is a more complex task compared to a conventional two-period two
sequence crossover trial. Secondly, the current standard for the analysis of replicate design trials is a
likelihood-based linear mixed model with random subject effects.
The question of whether to use fixed or random effects is not important for the standard two period,
two sequence (22) crossover trial. In section 4.1.8 of the guideline it is stated that subjects in a
crossover trial who do not provide evaluable data for both of the test and reference products should
not be included. Provided this is followed the confidence intervals for the formulation effect will be the
same regardless of whether fixed or random effects are used.
Therefore all that remains to be discussed is the analysis method for replicate designs. In section 2
three models for analysing data from replicate bioequivalence trials are considered. To illustrate these
approaches, in section 3 data from a four-period unbalanced study (see data set I) and data from a
three-period balanced study (data set II) were analysed using different statistical models and
computer programs.

2. Studied methods
2.1 Approach compatible with CHMP guideline (Method A)

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 20/47

The approach envisaged when the current guideline was written was to simply use the same analysis
method for replicate designs as is used for 22 trials.

proc glm dat a=r epl i cat e;
cl ass f or mul at i on subj ect per i od sequence;
model l ogDATA= sequence subj ect ( sequence) per i od f or mul at i on;
est i mat e " t est - r ef " f or mul at i on - 1+1;
t est h=sequence e=subj ect ( sequence) ;
l smeans f or mul at i on / adj ust =t pdi f f =cont r ol ( " R" ) CL al pha=0.10;
run;

For this model there is only one variance term estimated,
2
w,
the within subject variability.

2.2. Slight modification to approach compatible with CHMP guideline (Method B)
The same model as specified above could be used in PROC MIXED and subject specified as a random
effect.

proc mixed dat a=r epl i cat e;
cl ass f or mul at i on subj ect per i od sequence;
model l ogDATA= sequence per i od f or mul at i on;
r andomsubj ect ( sequence) ;
est i mat e " t est - r ef " f or mul at i on - 1 1 / CL al pha=0.10;
run;

This means there are two variance terms estimated
2
w
and
2
b
, as a distribution is also fitted to the
between subject variability. If subject is a fixed effect (as in the previous model) each subject is
treated as being selected in some way rather than being sampled from a random distribution and a
subject effect is estimated individually for each patient as is done for the period effect.
This model will give the same results as Method A if all subjects included in the analysis provide data
for all treatment periods.

2.3. Method C
The FDA Guidance for Industry document Statistical approaches to establishing bioequivalence
specifies the code to be used for the analysis of replicate designs using PROC MIXED.

proc mixed dat a=r epl i cat e;
cl asses sequence subj ect per i od f or mul at i on;
model l ogDATA= sequence per i od f or mul at i on / ddf m=sat t er t h;
r andomf or mul at i on/ t ype=FA0( 2) sub=subj ect G;
r epeat ed/ gr p=f or mul at i on sub=subj ect ;
est i mat e ' t est - r ef ' f or mul at i on - 1 1/ CL al pha=0.10;
run;

This model allows a different subject effect for each formulation (i.e. a subject by formulation
interaction), and therefore has 5 variance terms (within subject for reference, within subject for test,
between subject for test, between subject for reference, covariance for between subject test and
reference the last three are combined to give the subject formulation interaction variance
component.)


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 21/47

This model will provide the same point estimate as methods A and B if all subjects provide data for all
treatment periods. However it will generally give wider confidence intervals than those produced by
methods A and B.

3. Results
3.1. Data set I
The following data reflect a four period crossover study where subjects receive both test and reference
twice, with some subjects providing data for only a subset of the treatment periods. Results obtained
with methods A, B and C are shown in the following table.

Point
estimate
90% confidence
interval
Method A (guideline recommended) 115.66 107.11, 124.89
Method B (random effects) 115.73 107.17, 124.97
Method C (random effects with
interaction)
115.66 107.10, 124.89

Within subject CV% (from method C) reference 47.3%, test 35.3%
The results are generally very similar although missing treatment periods for some subjects causes the
results to be different for all three approaches.
3.2. Data set II
Data of a three period crossover study where all subjects receive reference twice and test once were
analyzed using Methods A, B and C.
The results are given in the Table below

Point
estimate
90% confidence
interval
Method A (guideline recommended) 102.26 97.32, 107.46
Method B (random effects) 102.26 97.32, 107.46
Method C (random effects with
interaction)
102.26 97.05, 107.76

Within subject CV% (from method C) reference 11.5%
As there are no subjects with missing treatment periods the results from methods A and B are
identical, and the point estimate is the same for all three approaches. Method C gives wider intervals.
3.3. Alternative computer programs
SAS (version 9.1, SAS Institute Inc, NC) was used in the previous computations. Results obtained by
alternative, validated statistical programs are also acceptable except spreadsheets because outputs of
spreadsheets are not suitable for secondary assessment.
3.4. Estimating the within subject variability
The guideline introduces the possibility of widening the acceptance limits for C
max
if the within-subject
variability for the reference product is greater than 30%. This is calculated using:

1 e 100 (%) CV
2
WR
s
=


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 22/47

The widening is on a smooth function, i.e. the permitted widening increases as the variability increases
(to a maximum of 50%). It is not an all or nothing criteria with 30% being a critical point.
An advantage of Method C is that it directly calculates s
2
wr
However, sometimes the algorithm fails to
converge. For that reason the preferred way to get an unbiased estimate of
2
wr
is using the data from
the reference product only.
The following code removes all the test data from the data-set and then fits a model where the
residual variance corresponds to the within subject variance for the test product.

data var ;
set r epl i cat e;
i f f or mul at i on=' R' ;
run;

proc glm dat a=var ;
cl ass subj ect per i od sequence;
model l ogDATA= sequence subj ect ( sequence) per i od;
run;

Results obtained with the different methods for Data Set I and II are summarised in the table below.
Reference within subject CV%
Model A/B Model C
Data set I 47.0% 47.3%
Data set II 11.2% 11.5%

The data shows that the variability estimates given by the two approaches are very similar for these
examples. There is no dependence on random effects mixed models to estimate within subject
variability for a formulation.

4. Discussion
The Guideline on the Investigation of Bioequivalence (CPMP/QWP/EWP/1401/98 Rev. 1) recommends
analysing bioequivalence studies using ANOVA and specifying all factors, including subject, as fixed
rather than random.
For a 22 crossover trial the confidence intervals for the formulation effect will be the same regardless
of whether fixed or random effects are used for subject.
For replicate designs the results from the two approaches will differ if there are subjects included in the
analysis who do not provide data for all treatment periods. Either approach is considered scientifically
acceptable, but for regulatory consistency it is considered desirable to see the same type of analysis
across all applications.
For multi-period studies other, more complex statistical models are possible. One of the possibilities is
to include a subject by formulation interaction term. Analysis of data currently available shows that the
subject by formulation interaction is negligible and therefore models without the interaction effect
adequately control the type I error. Thus the same statistical models can be used regardless of the
design.




Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 23/47

5. Conclusion
The Guideline on the Investigation of Bioequivalence (CPMP/QWP/EWP/1401/98 Rev. 1) recommends
analysing bioequivalence studies using ANOVA and specifying all factors, including subjects, as fixed
rather than random. The analysis presented above show that this approach (Method A) is feasible even
for unbalanced replicate design studies. The advantage of this approach is that it is straightforward and
that it appears to be software and software option independent. A simple linear mixed model, which
assumes identical within-subject variability (Method B), may be acceptable as long as results obtained
with the two methods do not lead to different regulatory decisions. However, in borderline cases and
when there are many included subjects who only provide data for a subset of the treatment periods,
additional analysis using method A might be required.
For highly-variable drugs it is recommended to estimate the within subject variance using data from
the reference formulation only.


ANNEX

Data set I
SUBJECT DATA FORMULATION PERIOD SEQUENCE logDATA
1 2285.96 R 1 BABA 7.734541
1 1955.82 T 2 BABA 7.578565
1 1345.94 R 3 BABA 7.204848
1 2856.24 T 4 BABA 7.957261
2 3151.72 T 1 ABAB 8.055704
2 2589.3 R 2 ABAB 7.859143
2 2992.94 T 3 ABAB 8.004011
2 2413.4 R 4 ABAB 7.788792
3 3264.74 T 1 ABAB 8.090935
3 3257.92 R 2 ABAB 8.088844
3 3100.54 T 3 ABAB 8.039332
3 3094.16 R 4 ABAB 8.037272
4 1206.36 T 1 ABAB 7.095363
4 1306.56 R 2 ABAB 7.175153
4 1583.12 T 3 ABAB 7.367153
4 1349.44 R 4 ABAB 7.207445
5 3880.9 R 1 BABA 8.263822
5 7322.88 T 2 BABA 8.898759
5 4429.66 R 3 BABA 8.396078
5 3322.88 T 4 BABA 8.108587
6 978.08 R 1 BABA 6.885591
6 1211.04 T 2 BABA 7.099235
6 973.88 R 3 BABA 6.881288
6 1150.8 T 4 BABA 7.048213
7 2924.06 T 1 ABAB 7.980728
7 2289.98 R 2 ABAB 7.736298
7 2494.28 T 3 ABAB 7.821755
7 3239.14 R 4 ABAB 8.083063
8 2425.46 R 1 BABA 7.793776
8 3705.74 T 2 BABA 8.217638
8 1891.06 R 3 BABA 7.544893
8 8979.12 T 4 BABA 9.102657
9 3825.02 R 1 BABA 8.249319
9 5315.04 T 2 BABA 8.578296
9 5813.16 R 3 BABA 8.667880

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 24/47

9 11475.9 T 4 BABA 9.348004
10 4112.26 R 1 BABA 8.321728
10 3822.86 T 2 BABA 8.248754
10 2459.82 R 3 BABA 7.807843
10 4616.76 T 4 BABA 8.437448
11 3170.3 T 1 ABAB 8.061581
11 2267.1 R 2 ABAB 7.726257
11 1703.32 R 4 ABAB 7.440335
12 2997.18 T 1 ABAB 8.005427
12 2954.78 R 2 ABAB 7.991179
12 5252.66 T 3 ABAB 8.566490
12 3744.54 R 4 ABAB 8.228054
13 2055.7 T 1 ABAB 7.628372
13 983.3 R 2 ABAB 6.890914
13 1771.3 T 3 ABAB 7.479469
13 3293.18 R 4 ABAB 8.099609
14 1590.62 R 1 BABA 7.371879
14 1141.54 T 2 BABA 7.040134
14 1238.34 R 3 BABA 7.121527
14 1285.8 T 4 BABA 7.159136
15 1470.5 T 1 ABAB 7.293358
15 1122.84 R 2 ABAB 7.023616
15 1592.18 T 3 ABAB 7.372859
15 1753.16 R 4 ABAB 7.469175
16 1886.14 R 1 BABA 7.542288
16 2077.28 T 2 BABA 7.638815
16 2197.62 R 3 BABA 7.695130
16 2194.64 T 4 BABA 7.693773
17 629.16 T 1 ABAB 6.444386
17 498.34 R 2 ABAB 6.211283
17 551.74 T 3 ABAB 6.313077
17 382.18 R 4 ABAB 5.945892
18 464.96 R 1 BABA 6.141951
18 2949.84 T 2 BABA 7.989506
18 1205.58 R 3 BABA 7.094716
18 2145.96 T 4 BABA 7.671342
19 1889.26 R 1 BABA 7.543940
19 5837.14 T 2 BABA 8.671996
19 2375.84 R 3 BABA 7.773106
19 1673.46 T 4 BABA 7.422649
20 793.44 T 1 ABAB 6.676378
20 1169.72 R 2 ABAB 7.064520
20 1072.8 R 4 ABAB 6.978027
21 2085.78 R 1 BABA 7.642898
21 2373.2 T 2 BABA 7.771995
21 1557 R 3 BABA 7.350516
21 2135.28 T 4 BABA 7.666353
22 288.06 R 1 BABA 5.663169
22 309.98 T 2 BABA 5.736508
22 324.18 R 3 BABA 5.781299
22 307.58 T 4 BABA 5.728735
23 524.8 T 1 ABAB 6.263017
23 372.84 R 2 ABAB 5.921149
23 518.92 T 3 ABAB 6.251750
23 604.56 R 4 ABAB 6.404501
24 5866.94 T 1 ABAB 8.677088

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 25/47

24 5547.78 T 3 ABAB 8.621153
24 4386.8 R 4 ABAB 8.386355
25 4008.46 T 1 ABAB 8.296162
25 1898.84 R 2 ABAB 7.548998
25 1565.22 T 3 ABAB 7.355782
25 4875.32 R 4 ABAB 8.491941
26 1197.46 T 1 ABAB 7.087958
26 330.82 R 2 ABAB 5.801574
26 1276.16 T 3 ABAB 7.151611
26 394.82 R 4 ABAB 5.978430
27 13823.18 R 1 BABA 9.534102
27 7618.82 T 2 BABA 8.938377
27 9493.34 R 3 BABA 9.158346
27 8928.44 T 4 BABA 9.096997
28 940.86 R 1 BABA 6.846794
28 1188.7 T 2 BABA 7.080616
28 882.02 R 3 BABA 6.782215
28 1226.38 T 4 BABA 7.111822
29 2175.24 R 1 BABA 7.684894
29 2654.36 T 2 BABA 7.883959
29 3235.26 R 3 BABA 8.081865
29 3033.3 T 4 BABA 8.017406
30 1194.9 T 1 ABAB 7.085818
30 826.66 R 2 ABAB 6.717393
30 610.38 T 3 ABAB 6.414082
30 594.14 R 4 ABAB 6.387115
31 4108.68 R 1 BABA 8.320857
31 7399.52 T 2 BABA 8.909170
31 4461.62 T 4 BABA 8.403267
32 792.22 T 1 ABAB 6.674839
32 999.74 R 2 ABAB 6.907495
32 1179.4 T 3 ABAB 7.072761
32 1678.96 R 4 ABAB 7.425930
33 3925.52 R 1 BABA 8.275254
33 3789.74 T 2 BABA 8.240053
33 3463.82 R 3 BABA 8.150127
33 4576.64 T 4 BABA 8.428720
34 1708.58 R 1 BABA 7.443418
34 2500.84 T 2 BABA 7.824382
34 1263.3 R 3 BABA 7.141483
34 2048.42 T 4 BABA 7.624824
35 943.06 T 1 ABAB 6.849130
35 769.22 R 2 ABAB 6.645377
35 848.8 T 3 ABAB 6.743824
35 1193.88 R 4 ABAB 7.084964
36 2540.42 T 1 ABAB 7.840085
36 2091.18 R 2 ABAB 7.645484
36 2583.66 T 3 ABAB 7.856962
36 1993.98 R 4 ABAB 7.597888
37 851.44 T 1 ABAB 6.746929
37 653.88 R 2 ABAB 6.482924
37 2371.3 T 3 ABAB 7.771194
37 1275.38 R 4 ABAB 7.150999
38 6054.76 R 1 BABA 8.708600
38 7322.18 T 2 BABA 8.898663
38 6746.98 R 3 BABA 8.816850

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 26/47

38 7130.7 T 4 BABA 8.872165
39 5825.64 T 1 ABAB 8.670024
39 6462.82 R 2 ABAB 8.773821
39 7400.48 T 3 ABAB 8.909300
39 6196.84 R 4 ABAB 8.731795
40 1690.42 R 1 BABA 7.432732
40 1292.9 T 2 BABA 7.164643
40 1522.4 R 3 BABA 7.328043
40 1066.58 T 4 BABA 6.972213
41 2783.06 R 1 BABA 7.931306
41 1149.08 T 2 BABA 7.046717
41 877.92 R 3 BABA 6.777555
41 572.42 T 4 BABA 6.349873
42 4759.06 T 1 ABAB 8.467805
42 5831.92 R 2 ABAB 8.671102
42 4154.76 R 4 ABAB 8.332010
43 5399.28 T 1 ABAB 8.594021
43 5425.9 R 2 ABAB 8.598939
43 4344.5 T 3 ABAB 8.376666
43 4507.04 R 4 ABAB 8.413396
44 5611.1 T 1 ABAB 8.632502
44 5444.14 R 2 ABAB 8.602295
44 4805.9 T 3 ABAB 8.477600
44 4960.66 R 4 ABAB 8.509294
45 707.68 R 1 BABA 6.561992
45 3681.66 T 2 BABA 8.211119
45 18454.26 R 3 BABA 9.823051
45 1003.46 T 4 BABA 6.911209
46 2400.64 T 1 ABAB 7.783491
46 1420.6 R 2 ABAB 7.258835
46 1146.68 T 3 ABAB 7.044626
46 5005.72 R 4 ABAB 8.518337
47 483.08 R 1 BABA 6.180182
47 1033.3 T 2 BABA 6.940513
47 644.54 R 3 BABA 6.468537
47 675.3 T 4 BABA 6.515157
48 2157.08 R 1 BABA 7.676511
48 3117.36 T 2 BABA 8.044742
48 2816.14 R 3 BABA 7.943122
48 2850.4 T 4 BABA 7.955215
49 14261.54 T 1 ABAB 9.565322
49 26489.56 R 2 ABAB 10.184506
49 23525.66 T 3 ABAB 10.065847
49 21243.76 R 4 ABAB 9.963818
50 1552.24 T 1 ABAB 7.347454
50 1569.32 R 2 ABAB 7.358398
50 2090 T 3 ABAB 7.644919
50 1479.98 R 4 ABAB 7.299784
51 3834.44 R 1 BABA 8.251779
51 4899.76 T 2 BABA 8.496942
51 3702.9 R 3 BABA 8.216872
51 5677.02 T 4 BABA 8.644182
52 5925.92 R 1 BABA 8.687091
52 967.9 T 2 BABA 6.875129
52 797.02 R 3 BABA 6.680880
52 939.38 T 4 BABA 6.845220

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 27/47

53 3528.48 T 1 ABAB 8.168622
53 2037.36 R 2 ABAB 7.619410
53 3211.68 T 3 ABAB 8.074549
53 2906.74 R 4 ABAB 7.974787
54 937.16 R 1 BABA 6.842854
54 6327.96 T 2 BABA 8.752733
54 1054.92 R 3 BABA 6.961220
54 1766.02 T 4 BABA 7.476484
55 3437.98 T 1 ABAB 8.142639
55 3731.8 R 2 ABAB 8.224646
55 4832.72 T 3 ABAB 8.483165
55 3310.24 R 4 ABAB 8.104776
56 1011.14 T 1 ABAB 6.918834
56 654.02 R 2 ABAB 6.483138
56 858.58 T 3 ABAB 6.755280
56 908.12 R 4 ABAB 6.811377
57 1003.34 R 1 BABA 6.911090
57 4739.94 T 2 BABA 8.463780
57 697.84 R 3 BABA 6.547990
57 2504.52 T 4 BABA 7.825852
58 6496.34 R 1 BABA 8.778994
58 5949.36 T 2 BABA 8.691039
58 6003.38 R 3 BABA 8.700078
58 6373.72 T 4 BABA 8.759939
59 1247.58 R 1 BABA 7.128961
59 1116.88 T 2 BABA 7.018294
59 1166.74 R 3 BABA 7.061969
59 2658.38 T 4 BABA 7.885472
60 33929.62 T 1 ABAB 10.432044
60 24943.44 R 2 ABAB 10.124366
60 19110.22 T 3 ABAB 9.857979
60 12805.18 R 4 ABAB 9.457605
62 2280.5 T 1 ABAB 7.732150
62 1714.48 R 2 ABAB 7.446865
62 4034.28 T 3 ABAB 8.302583
62 3420.76 R 4 ABAB 8.137618
63 3376.72 T 1 ABAB 8.124660
63 2242.8 R 2 ABAB 7.715480
63 1719.54 T 3 ABAB 7.449812
63 2342.32 R 4 ABAB 7.758897
64 912.34 R 1 BABA 6.816013
64 2104.42 T 2 BABA 7.651795
64 2061.04 R 3 BABA 7.630966
64 1496.5 T 4 BABA 7.310884
65 3957.94 R 1 BABA 8.283479
65 5895.6 T 2 BABA 8.681962
65 5859.58 R 3 BABA 8.675833
65 5073.48 T 4 BABA 8.531782
66 1165.7 T 1 ABAB 7.061077
66 1248.62 R 2 ABAB 7.129794
66 1168.68 T 3 ABAB 7.063630
66 1300.42 R 4 ABAB 7.170443
67 1197.4 R 1 BABA 7.087908
67 1119.34 T 2 BABA 7.020495
68 1709.72 R 1 BABA 7.444085
68 2532.4 T 2 BABA 7.836923

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 28/47

68 1581.02 R 3 BABA 7.365825
68 2807.4 T 4 BABA 7.940014
69 2798.84 T 1 ABAB 7.936960
69 2454.1 R 2 ABAB 7.805515
69 5334.84 R 4 ABAB 8.582014
70 4318.42 R 1 BABA 8.370645
70 2182.66 T 2 BABA 7.688300
70 1649.16 R 3 BABA 7.408021
70 1620.32 T 4 BABA 7.390379
71 470.24 T 1 ABAB 6.153243
71 208.04 R 2 ABAB 5.337730
72 2098.3 T 1 ABAB 7.648883
72 1919.76 R 2 ABAB 7.559955
72 2817.76 T 3 ABAB 7.943698
72 2041 R 4 ABAB 7.621195
73 6667.32 T 1 ABAB 8.804973
73 5289.84 R 2 ABAB 8.573543
73 7300.28 T 3 ABAB 8.895668
73 9711.84 R 4 ABAB 9.181101
74 2036.76 R 1 BABA 7.619116
74 1948.04 T 2 BABA 7.574579
74 1539.58 R 3 BABA 7.339265
74 2079.14 T 4 BABA 7.639710
75 767.3 T 1 ABAB 6.642878
75 1046.3 R 2 ABAB 6.953015
75 1390.36 T 3 ABAB 7.237318
75 3019.18 R 4 ABAB 8.012741
76 12097.5 T 1 ABAB 9.400754
76 12694.42 R 2 ABAB 9.448918
76 10999.24 T 3 ABAB 9.305581
76 9406.52 R 4 ABAB 9.149158
77 1115.5 R 1 BABA 7.017058
77 1115.3 T 2 BABA 7.016879
77 1111.78 R 3 BABA 7.013718
77 2352.82 T 4 BABA 7.763370
78 20373.54 R 1 BABA 9.921992
78 13689.6 T 2 BABA 9.524392
78 20585.02 R 3 BABA 9.932319
78 24498.14 T 4 BABA 10.106352

Data Set II
SUBJECT DATA FORMULATION PERIOD SEQUENCE logDATA
1 4053.6 R 1 2 8.307361
1 3970.4 T 2 2 8.286622
1 3748.8 R 3 2 8.229191
2 2986.2 R 1 2 8.001757
2 2378.8 T 2 2 7.774351
2 2804.6 R 3 2 7.939016
3 3464.4 R 1 3 8.150295
3 3340.2 R 2 3 8.113786
3 4028.8 T 3 3 8.301224
4 4105 T 1 1 8.319961
4 3191.2 R 2 1 8.068152
4 3803.6 R 3 1 8.243703
5 4767.8 T 1 1 8.469640
5 4542.6 R 2 1 8.421255

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 29/47

5 3940 R 3 1 8.278936
6 3050.8 R 1 3 8.023159
6 3027.2 R 2 3 8.015393
6 2419.6 T 3 3 7.791358
7 2530.2 R 1 2 7.836054
7 3072 T 2 2 8.030084
7 2962.6 R 3 2 7.993823
8 2205 T 1 1 7.698483
8 2041.4 R 2 1 7.621391
8 2018 R 3 1 7.609862
9 4647.6 R 1 2 8.444106
9 4159.6 T 2 2 8.333174
9 3400 R 3 2 8.131531
10 2228.2 T 1 1 7.708949
10 2360.4 R 2 1 7.766586
10 2221.2 R 3 1 7.705803
11 1863.8 R 1 3 7.530373
11 2212.4 R 2 3 7.701833
11 2394.4 T 3 3 7.780888
12 2278.4 R 1 3 7.731229
12 3170.4 R 2 3 8.061613
12 3927.2 T 3 3 8.275682
13 2640.4 R 1 3 7.878686
13 2430.4 R 2 3 7.795811
13 2869.2 T 3 3 7.961789
14 3030.8 R 1 2 8.016582
14 2459.8 T 2 2 7.807835
14 2970.4 R 3 2 7.996452
15 2254.4 R 1 2 7.720639
15 1994.8 T 2 2 7.598299
15 2724.4 R 3 2 7.910003
16 2959.6 T 1 1 7.992809
16 3442 R 2 1 8.143808
16 3342.6 R 3 1 8.114504
17 2396.8 T 1 1 7.781890
17 2659.4 R 2 1 7.885856
17 2172 R 3 1 7.683404
18 2725 R 1 3 7.910224
18 2805.6 R 2 3 7.939373
18 3146.6 T 3 3 8.054078
19 2418.8 R 1 2 7.791027
19 2749.8 T 2 2 7.919283
19 2504 R 3 2 7.825645
20 2662.4 R 1 3 7.886983
20 2929.8 R 2 3 7.982689
20 3037.2 T 3 3 8.018691
21 2869.6 R 1 3 7.961928
21 2666.4 R 2 3 7.888485
21 3069 T 3 3 8.029107
22 2949 T 1 1 7.989221
22 2926.8 R 2 1 7.981665
22 2855.4 R 3 1 7.956967
23 3154.8 T 1 1 8.056680
23 3185.6 R 2 1 8.066396
23 3548.6 R 3 1 8.174308
24 1874.8 R 1 2 7.536257

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 30/47

24 1808.8 T 2 2 7.500419
24 2730.8 R 3 2 7.912350



Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 31/47

11. Requirements for demonstration of bioequivalence for generics of
biphasic modified release formulations for oral use
Biphasic modified release formulations are characterised by two phases of drug release: a first phase
determined by the immediate release dose fraction to provide a therapeutic drug level shortly after
administration, and a second extended release phase to provide the dose fraction required to maintain
an effective therapeutic level for a prolonged period. The clinical rationale behind their development is
that a rapid onset of action is required in addition to subsequent prolonged release characteristics.
The principle criteria for the establishment of bioequivalence as stated in the Note for guidance on
modified release oral and transdermal dosage forms, Section II (CPMP/EWP/280/96) apply also for
multiphasic modified release products. Pharmacokinetic parameters for the establishment of
bioequivalence of a generic biphasic modified release formulations with the corresponding innovator
formulation need to characterise both phases of drug release to ensure that the plasma concentration
time curves for the generic product correspond to those for the reference product. Therefore,
parameters need to establish the equivalence in terms of early drug absorption, reflecting mostly the
immediate release component of the product, as well as the equivalence in terms of the second phase
reflecting the prolonged release component.
For the pharmacokinetic evaluation, these two phases should be separated through a cut-off time
point, which needs to be pre-specified and universally applied to all subjects and for both test product
and reference product. The identification of this cut-off time point should aim to describe the plasma
concentrations in the first phase driven by the immediate release dose fraction whilst avoiding bias
through an increasing contribution of the extended release phase.
Equivalence needs to be shown for both extent and rate of absorption (reflecting AUC and C
max
for
conventional bioequivalence criteria), separately for both phases:
For the first phase, the assessment of equivalence should be based on the truncated AUC from
t=0 until the cut-off time describing the immediate release dose fraction, and on Cmax during
the first phase.
For the second phase, the assessment of equivalence should be based on the AUC from the
cut-off time until the end of observation period, and on Cmax during the second phase.
These considerations are in principle valid for studies in fed state and in fasting state. If no significantly
different pharmacokinetic profile between fasting and fed state is expected then the cut-off time point
should be identical.
These recommendations are based on the present Note for guidance on modified release oral and
transdermal dosage forms, Section II (CPMP/EWP/280/96) and represent the current regulatory
thinking. It should be noted that this guideline is currently under revision hence recommendations
regarding these aspects may change in the revised guideline.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 32/47

12. Effect of sorbitol on the pharmacokinetics of highly permeable drug
substances
The CMDh asked for a view on the extent to which the results reported by Chen et al. (1) regarding the
effect of sorbitol on bioavailability of metoprolol, taken together with relevant regulatory experience
regarding the influence of sorbitol on the oral bioavailability of drug substances, are applicable to other
highly permeable drug substances (BCS class 1 and 2).
There is scarce information in the literature (1-5) regarding the effect of sorbitol on the absorption of
BCS class I and II (highly permeable drug substances). The article by Chen et al (1) (showing no effect
on metoprolol absorption) and another one by Fassihi (2) (showing no effect on Cmax or AUC but an
effect on Tmax of theophylline upon 10 g of sorbitol) are worth mentioning.
In Chen et als article (1), the effect of sorbitol on the absorption of metoprolol (BCS class I) and
ranitidine (BCS class III) has been studied. No significant effect of sorbitol (5 g) on the extent (AUC)
and a 23% reduction in rate (Cmax) of absorption of a single dose of metoprolol has been recorded,
whereas a significant effect has been observed on both AUC and Cmax (44% and 51% reduction,
respectively) when sorbitol (5 g) and ranitidine (BCS class III) were administered concomitantly. From
these data, the best estimate of a single dose threshold for the sorbitol effect on drug bioavailability is
probably around 1 g, affecting all drug BCS classes but mainly low permeability drug substances.
Therefore there is no straightforward answer to this question until more data is collected to determine
the actual threshold by exploring sorbitol doses lower than 1.25 g.
The putative effect of sorbitol on GI physiology affecting drug absorption is generally accepted to
derive from its osmotic effect, accelerating intestinal transit and increasing intestinal water content.
The first effect suggests a higher impact on the absorption of low permeability drugs. The latter can
lower the diffusion driving force due to dilution, affecting all drug BCS classes.
Therefore any correlation of sorbitol absorption effect with solubility or permeability is in principle
difficult to establish.
It also needs to be recognized that sorbitol intolerance is largely described in the literature (6, 7). This
means that a dose effect relationship cannot be established universally due to individual susceptibility.
Even minute amounts of sorbitol can elicit a GI effect in a sub-population.
Consistently with these results, the Bioequivalence Guideline (CPMP/EWP/QWP/1401/98 Rev. 1) states
in Appendix II, Oral solutions:
If the test product is an aqueous oral solution at time of administration and contains an active
substance in the same concentration as an approved oral solution, bioequivalence studies may be
waived. However if the excipients may affect gastrointestinal transit (e.g. sorbitol, mannitol, etc.), [],
a bioequivalence study should be conducted, unless the differences in the amounts of these excipients
can be adequately justified by reference to other data. The same requirements for similarity in
excipients apply for oral solutions as for Biowaivers (see Appendix III, Section IV.2 Excipients.
Further recommendations in Appendix III, section IV.2 on excipients state: As a general rule, for both
BCS-class I and III drug substances [] Excipients that might affect bioavailability should be
qualitatively and quantitatively the same in the test product and the reference product.
Therefore, strict compliance with the Bioequivalence Guideline is recommended to be followed in the
development and assessment of generic applications.
Sorbitol intolerance should be taken into consideration in the labeling of sorbitol containing drug
products.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 33/47


References
1. Chen, M., Straughn, A., Sadrieh, N. et al (2007). A Modern View of Excipient Effects on
Bioequivalence: Case Study of Sorbitol. Pharm. Res. 24, 73-80.
2. R. Fassihi, R. Dowse, and S. S. D. Robertson (1991). Influence of Sorbitol Solution on the
Bioavailability of Theophylline. Int. J. Pharm. 72:175-178
3. D. A. Adkin, S. S. Davis, R. A. Sparrow, P. D. Huckle, A. J. Philips, and I. R. Wilding (1995). The
Effects of Pharmaceutical Excipients on Small Intestinal Transit. Br. J. Clin. Pharmacol. 39:381-387
4. D. A. Adkin, S. S. Davis, R. A. Sparrow, P. D. Huckle, and I. R. Wilding (1995). The Effect of
Mannitol on the Oral Bioavailability of Cimetidine. J. Pharm. Sci. 84:1405-1409
5. S. van Os, M. Relleke and Muniz Piniella (2007) Lack of Bioequivalence between Generic
Risperidone Oral Solution and Originator Risperidone Tablets, Int. J. Clin. Pharmacol., 45: 293-299
6. Born P., The clinical impact of carbohydrate malabsorption (2011) Arab J Gastroenterol.
Mar;12(1):1-4. Epub 2011 Feb 5.
7. Fernndez-Baares F, Esteve M, Viver JM. (2009) Fructose-sorbitol malabsorption. Curr
Gastroenterol Rep. Oct;11(5):368-74.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 34/47

13. Requirement to perform incurred sample reanalysis
The requirement to perform incurred sample reanalysis (ISR) has been introduced with the Guideline
on bioanalytical method validation (EMEA/CHMP/EWP/192217/2009). Given that this is a new
regulatory requirement with the need for consistency in its introduction, the PKWP was asked to
discuss if it is possible to give recommendations on how the absence of ISR should be handled and
whether it is possible to identify other factors which could be assessed in the absence of ISR to support
the validity of the analytical method.
Introduction
Incurred sample reanalysis (ISR) is applied to assess the reliability of bioanalytical methods used in
pre-clinical toxicokinetic studies and for a variety of clinical pharmacology studies including
bioavailability, bioequivalence, pharmacokinetic, interaction and comparability studies. The need for
incurred sample reanalysis is discussed already since 2006
2
and regulators supported the need for
incurred sample reanalysis also considering significant bioanalytical deficiencies observed in studies.
Therefore, although incurred sample reanalysis is a requirement introduced in Europe for the first time
with the new EMA Guideline on bioanalytical method validation (EMEA/CHMP/EWP/192217/2009),
which came into force in February 2012, it should be noted that the scientific need to perform ISR as
an element of bioanalytical method validation was already identified much earlier. ISR should
therefore be considered as part of the validation of the analytical method during study sample analysis.
Different sources can be identified which might contribute to the failure of ISR. Some sources may be
more likely to occur than other depending on the method, active substance, and analyst, however they
cannot be excluded. Sources of ISR failure may be:
Execution, i.e. switched samples, instrument issues, scientist performance of method
Method, i.e. metabolite interferences, back conversion of metabolites, poor ruggedness,
internal standard response
Samples, i.e. matrix effects, mislabelling, handling
It is recognized that some of these sources are also likely to occur during validation, like switching
samples and mislabelling.
ISR failure and thus lack of the reliability of the study outcome can happen in each study and as such
it is difficult to generalise it. Especially with pivotal studies it should be ensured that the results are
reliable. However it is also understood that ISR is an additional confirmation of results next to a
complete validation.
Introduction of ISR as a regulatory requirement
The principles for the implementation of a guideline are outlined in the Procedure for European Union
guidelines and related documents within the pharmaceutical legislative framework
(EMEA/P/24143/2004 Rev.1). While applicants may, with the agreement of the competent authority
concerned, choose to apply a guideline in advance of the date for coming into operation of a guideline,
competent authorities should await this date before requiring a guideline to be taken into account for
assessments. The Guideline on bioanalytical method validation came into force on 1 February 2012
2
Viswanathan CT, Bansal S, Booth B et al. Workshop/Conference Report: Quantitative bioanalytical methods
validation and implementation: best practices for chromatographic and ligand binding assays. AAPS J. 9(1), E30
E42 (2007)

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 35/47


meaning that as of this date this document sets the applicable requirements for the regulatory review
of applications.
It is acknowledged in the above-mentioned principles that in some circumstances it may not be
possible for applicants to fully comply with new guidelines within this timeframe (e.g. data generated
from trials started before the implementation of the new guideline). In such cases, the applicant should
consider whether departure from the new guideline could be justified. The applicant's justification will
then be considered on a case-by-case basis by the relevant competent regulatory authorities.
In compliance with this framework, the regulatory assessment requires the review of the bioanalytical
method validation in any application against the current regulatory standards as set out in the
guideline, including the requirement to address incurred sample reanalysis. If an element of the
validation is missing, e.g. lack of incurred sample reanalysis, then this would need to be scientifically
justified by the applicant. Such justification can be considered in the framework of the above exception
that a particular validation has been performed before the bioanalytical guideline came into force, i.e.
February 2012. Any justification will need to be reviewed on a case-by-case basis considering the
overall validation data, the study results, as well as the reliance of the application on these data.
Considerations regarding a potential justification for the lack of ISR data
The attempt to scientifically justify the lack of ISR is considered only appropriate for the very practical
reason that a study was performed before the guideline on bioanalytical method validation came into
force.
For the scientific justification of the lack of ISR the applicant should take all the following points into
consideration:
metabolite back conversion:
the applicant should support that back conversion is not an issue for the drug compound or
that the risk of back conversion on the outcome of the study results is low as for instance it is
known that the drug compound is (almost) not metabolised. For drug compounds for which it is
known that back conversion is an issue, i.e. clopidogrel, atorvastatin, ramipril, lack of ISR is
considered not acceptable.
other ISR data obtained in the same laboratory:
ISR data obtained for the same analyte from other studies carried out in the same laboratory
and with the same analytical method may be used as supportive data to justify the lack of ISR.
data from repeat analysis:
In most studies repeat analysis of study samples has to be carried out for different reasons.
Repeat analysis can be considered as ISR in certain situations, however due to the nature of
the reanalysis (for instance run acceptance criteria failure) those data are considered not
reliable. The applicant should report the data of these reanalysis and take into account and
discuss the reason for the reanalysis in the justification for supportive data.
In case of a multi analyte analysis, if the repeat analysis was due to run acceptance criteria
failure for one of the analytes, but the other has passed, the results of the analyte(s) which
passed can be used to infer ISR, if analysed.
the obtained pharmacokinetic data in the study:
the applicant should compare the obtained pharmacokinetic data with data obtained previously
or with reported data and should show that these are comparable

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 36/47

90% confidence interval:
as one element of such justification, if applicable, the applicant could also take into
consideration the width of the 90% confidence interval and the ratio to possibly justify that a
false positive outcome due to ISR problems has a low probability.
The last two bullet points need to be thoroughly discussed specifically for bioequivalence studies.
The applicant should also consider the overall reliance of the application on the data generated with
the bioanalytical method in question. For new molecular entities the pivotal basis of the application
normally rests on clinical efficacy and safety studies, nevertheless pharmacokinetic studies in such an
application provide significant information (e.g. general pharmacokinetic profile, interactions), which is
also reflected in the labelling, hence the validity of such data needs to be sufficiently ensured. Abridged
applications may exclusively rely on pharmacokinetic data, e.g. bioequivalence studies, making overall
validity of these data paramount. Therefore, the validity of the data needs to be considered for the
assessment of the application and the specific study considering whether the data are pivotal or
supportive.
Conclusion
ISR is considered an element of the validation of the analytical method during study sample analysis.
It has been discussed for many years in the scientific community and recently been introduced as
regulatory requirement in the European guideline. Like for any deviation from a guideline requirement,
the lack of ISR requires a scientific justification by the applicant. Such justification could be considered
for validations which have been performed before the new guideline came into force. Its scientific
validity will need to be reviewed on a case-by-case basis in the light of the overall validation data, the
study outcome, as well as the reliance of the application on these data.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 37/47

14. Number of subjects in a two-stage bioequivalence study design
According to the Guideline on the Investigation of Bioequivalence (CPMP/QWP/EWP/1401/98 Rev.1), it
is acceptable to use a two-stage approach when attempting to demonstrate bioequivalence. The
question was raised whether there was a minimum number of subjects that should be included in the
second stage of such a design.
Discussion
From the perspective of type I error control it is considered that there is no minimal number of
subjects to be included in the second stage of a two-stage design, so long as it can be demonstrated
that the type I error of the study is controlled. However, the analysis model for analysing the combined
data also needs to be considered.
The CHMP guideline on the investigation of bioequivalence (Doc. Ref.: CPMP/EWP/QWP/1401/98 Rev.
1/ Corr) states: When analysing the combined data from the two stages, a term for stage should be
included in the ANOVA model. In addition, to account for the fact that the periods in the first stage are
different from the periods in the second stage, a term for period within stage is required. Therefore,
the expected ANOVA model for analysis of the combined data from a two-stage design would have the
following terms: stage, sequence, sequence*stage, subject (sequence*stage), period (stage),
formulation. To fit this model it is necessary to have in each stage at least one patient in each
sequence so a minimum of two patients in each stage of the study, but more if both happen to be
randomised to the same sequence.
A model which also includes a term for a formulation*stage interaction would give equal weight to the
two stages, even if the number of subjects in each stage is very different. The results can be very
misleading hence such a model is not considered acceptable. Furthermore, this model assumes that
the formulation effect is truly different in each stage. If such an assumption were true there is no
single formulation effect that can be applied to the general population, and the estimate from the
study has no real meaning.
Conclusion
1) The expected analysis for the combined data in a two-stage design is ANOVA with terms for stage,
sequence, sequence*stage, subject (sequence*stage), period (stage), formulation.
2) This model can be fitted provided that in each stage, there is at least one subject randomised to
each sequence. This does not supersede the requirement for at least 12 subjects overall.
3) A term for a formulation*stage interaction should not be fitted.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 38/47

15. Bioequivalence studies for generic application of omega 3 fatty acid
ethylesters in a soft gelatine capsule
The CMDh asked for PKWPs view on a generic application of Omega-3 fatty acid ethylesters in a soft
gelatine capsule.
The capsule filling of both the generic and the innovator product comprised 1000 mg of the liquid
active substance, (omega 3 fatty acid ethylesters), without any excipients. The active substance fully
complied with the Ph Eur Monograph on Omega-3 fatty acid ethylesters (EE) which describes an active
substance including an allowed (although not defined) low amount of preservative.
Hence, the gelatin capsules only included the oily, liquid active substance, (omega 3 fatty acid
ethylesters). However, the liquid active substance contains a slightly different amount of preservative
alpha-tocopherol (as 70% in vegetable oil). Furthermore the composition of the capsule itself was
roughly the same as for the innovator product but with a slight difference in the amount of glycerol.
This particular situation is not addressed in the current guideline on the investigation of bioequivalence
(CPMP/EWP/QWP/1401/98 Rev. 1/ Corr **), i.e. a generic application referring to an oily liquid
composition in a soft gelatine capsule. Hence, the CMDh asked for PKWPs view on:
Whether a biowaiver would be acceptable in this specific type of drug formulation if fast and
comparable disintegration of the capsules has been demonstrated over the whole physiological
range (pH 1 6.8).
Should a bioequivalence trial be required, what would be the preferred study design (fed or
fasted).
In the case of fasted state conditions, would it be possible to determine bioequivalence
between drug products including in the analysis subjects that have presented erratic
absorption profiles, for which the extrapolation AUC
t-inf
could not be estimated or was >20% in
more than 50% of the subjects.
Discussion
Bioequivalence (BE) is a means to detect potential formulation differences between generics and
innovators. This implies that formulation differences are expected due to e.g. different excipients
(quantitatively and/or qualitatively) and/or different manufacturing processes.
Since the oily content of both capsule products including an allowed amount of preservative is
considered the active substance (PhEur monograph), a different formulation effect cannot be assumed.
Hence, requesting in vivo BE between test and reference could hardly be justified as both capsules
would contain the same amount of actives within accepted limits of variability without excipients
potentially causing different formulation effects. The possibility of different amounts of impurities is
expected to be controlled via the monograph, i.e. this could not be the reason for a BE study as it
refers to the active substance rather than the formulation.
Therefore, simple characterisation of capsule quality by comparative disintegration tests is deemed
sufficient. It should however be noted that the disintegration of capsule shells cannot be used as a BE
tool as such as it has no relation to any in vivo parameter, but simply describes capsule quality.
In summary, a biowaiver would be acceptable in this specific type of drug formulation if fast and
comparable disintegration of the capsules has been demonstrated over the whole physiological range
(pH 1 6.8). Since the liquid oily active substance of the capsules filled with omega-3 fatty acid EEs

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 39/47

will be directly available for absorption after rupture and disintegration and a different formulation
effect cannot be expected from the allowed preservative, in vivo BE study could be waived.
Should in vivo BE trial be requested, it should be performed under fed conditions for the following
reasons:
Plasma concentrations are markedly higher under fed conditions than those quantified in the
fasted state,
Plasma concentrations in the fasted state are rather low and erratic. Unreasonably low values
within the PK profiles render them invalid as they indicate the measurements of physiological
processes rather than pharmacokinetics.
The last point was addressed in the paragraph above. However, since this is considered a general
question not particularly related to the omega-3 fatty acid ethylesters in a soft gelatine capsule, it is
further discussed below.
Subjects for which erratic absorption prevent the calculation of extrapolated AUC and/or for which the
residual area is more than 20 % should still be included in the regular calculations and evaluation of
AUC
t
since this is the most relevant pharmacokinetic parameter to compare extent of absorption (see
section 4.1.8 in the guideline on the investigation of bioequivalence (CPMP/EWP/QWP/1401/98 Rev. 1/
Corr **)). However, the cited guideline clearly states that when this is true in more than 20 % of the
observations then the validity of the study may need to be discussed (see section 4.1.8 Evaluation;
Reasons for exclusion). Hence, only in exceptional cases it could still be possible to accept an
extrapolation larger than 20% in a significant number of subjects (>20% of the subjects concentration
- time profiles) if it is justified that AUC
t
has been calculated reliably and it is representative of the
extent of drug absorption from the products under comparison. Of note, this rule and reasoning does
not apply if the sampling period is 72h or more and AUC
0-72h
is used instead of AUC
t
.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 40/47

16. Acceptability of an additional strengths biowaiver when
bioequivalence to the reference product has been established with a
BCS-based biowaiver
The Guideline on the Investigation of Bioequivalence (CPMP/EWP/QWP/1401/98 Rev. 1/ Corr) states
that: If bioequivalence has been demonstrated at the strength(s) that are most sensitive to detect a
potential difference between products, in vivo bioequivalence studies for the other strength(s) can be
waived.
The PKWP was asked to comment on the acceptability of this approach when the bioequivalence of the
reference strength to the reference product has been investigated using the BCS (Biopharmaceutics
Classification System)-based biowaiver approach i.e., without an in vivo bioequivalence study.
Discussion
Bioequivalence is in principle demonstrated by means of in vivo bioavailability studies. These in vivo
studies can be waived if the product fulfils the requirements defined in surrogate tests like the BCS
biowaiver approach.
This is in accordance with the Guideline on the Investigation of Bioequivalence
(CPMP/EWP/QWP/1401/98 Rev. 1/ Corr) which states in this respect that: The BCS (Biopharmaceutics
Classification System)-based biowaiver approach is meant to reduce in vivo bioequivalence studies,
i.e., it may represent a surrogate for in vivo bioequivalence. In vivo bioequivalence studies may be
exempted if an assumption of equivalence in in vivo performance can be justified by satisfactory in
vitro data.
An additional strength biowaiver is a waiver designed to avoid repeating the same in vivo study at the
other strength level. Hence, when the Guideline on the Investigation of Bioequivalence
(CPMP/EWP/QWP/1401/98 Rev. 1/ Corr) states that: If bioequivalence has been demonstrated at the
strength(s) that are most sensitive to detect a potential difference between products, in vivo
bioequivalence studies for the other strength(s) can be waived, this implies that when bioequivalence
has been demonstrated in vivo for the test product, in vivo bioequivalence studies for the other
strength can be waived.
Indeed, the reference in the sentence above to the sensitivity to detect differences between test and
reference products only makes sense in the case of in vivo comparisons. This sensitivity varies
depending on the solubility and the pharmacokinetic linearity. In the case of highly soluble drugs, the
only drugs for which a BCS biowaiver is acceptable, the sensitivity to detect differences in vitro is the
same at all strengths. Thus, the reference to higher sensitivity at the highest strength refers to in vivo
studies. Further, the different sensitivities arising from non-linear pharmacokinetics only apply to in
vivo studies. Therefore, the intent of this text was to refer to in vivo studies as evidence of
bioequivalence.
Summary
Biowaiver of additional strength should be applied only when the test product have shown
bioequivalence to the reference product by means of an in vivo bioequivalence study.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 41/47

17. Question on a generic application for Quetiapine Lambda 200, 300,
400 mg prolonged release tablets
The CMDh consulted the PKWP for their input on a generic application (article 10.1 of Directive
2001/83/EC) for Quetiapine Lambda 200, 300, 400 mg prolonged release tablets. The originator
products strengths were 200 mg, 300 mg, 400 mg, prolonged release tablets.
The clinical development plan for Quetiapine Lambda 200, 300, 400 mg prolonged release tablets
consisted of a single-dose study under fasting and fed conditions with 200 mg strength in healthy
volunteers and a multiple-dose study with the highest, 400 mg tablet in schizophrenic patients.
The application for the 300 and 400 mg strength was referred to the CMDh. The PKWP input was
sought on the following points:
1/ Clinical development plan: the need for single dose bioequivalence studies in all strengths, where
single-dose study under fasting and fed conditions with 200 mg strength in healthy volunteers and a
multiple-dose study with the highest, 400 mg tablet in schizophrenic patients have shown
bioequivalence,
2/ The need for inclusion of early time points in the calculation of f2 values for a prolonged release
tablet in in-vitro dissolution data supportive of a biowaiver.
Preamble
The PKWP acknowledged the following limitations:
Single dose studies with doses higher than 200 mg are not feasible in healthy volunteers due
to unacceptably severe adverse effects,
Multiple dose studies with doses equal to or higher than 200 mg are not feasible in healthy
volunteers due to unacceptably severe adverse effects,
Single dose studies in patients are not feasible due to ethical reasons (interruption of
treatment).
Hence, the PKWPs feedback was based on the assumption that it was not possible to conduct the
study with the 300mg dose.
Discussion
1. Would a multiple dose study in the highest strength be considered sufficient to
demonstrate bioequivalence despite differences in the dissolution profiles, in case where a
single- dose study can be waived because of safety reasons?
PKWP response
In the case of Quetiapine Lambda the following statement from the MR NfG (1), applies:
In case of prolonged release single unit formulations with multiple strengths, a single dose study under
fasting conditions is required for each strength. Studies at steady state may be conducted with the
highest strength only if the same criteria for extrapolating bioequivalence studies are fulfilled as
described in the Note for Guidance for immediate release forms (linear pharmacokinetics, same
qualitative composition, etc.).
Therefore, the following is required:

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 42/47

1. Waive multiple dose studies for the 200 mg and 300 mg strengths based on conditions
applicable to IR forms as per BE GL currently in force (2). All conditions were fulfilled except
for dissolution (see below).
2. Waive single dose studies for the 300 mg and 400 mg studies based on exceptional
circumstances: single dose studies are not feasible both in healthy volunteers and patients
(see above). In this case the same rules for waiving different strengths should apply.
As a consequence, the only outstanding issue was the comparison of dissolution profiles.
Overall the dissolution data raised doubts on the extrapolation of the BE results only from the 400 mg
and the 200 mg strengths because the comparison of 200 mg vs. 400 mg at pH 4.5 and 6.8 does not
meet the f2 criterion. On the contrary, bioequivalent (BE) results could be extrapolated to the 300 mg
strength on the basis of dissolution data since respective comparisons complied with the f2 criterion.
It was then investigated whether the differences in the dissolution data were due to an active
substance effect (as a result of lack of sink conditions) or a formulation effect. As for the lack of sink
conditions, the results of a comparison of equivalent strengths of the test product (TP) (2X200 mg vs.
1X400 mg) at pH 4.5 and 6.8 suggested that the noncompliant results could be explained by an active
substance effect, not by a formulation effect. However, the results of a comparison of the 200 mg
strength of the reference product (RP) the 400 mg strength of the RP at pH 4.5 and 6.8 did not
suggest an active substance effect.
Given the exceptional circumstances that the single dose studies cannot be conducted in patients and
that the studies with doses higher than 200 mg cannot be conducted in healthy volunteers, only a
multiple 200 mg dose study in patients could have clarified these findings. . However, this study would
not be ethically acceptable since there was direct evidence that the lack of comparability between 200
mg and 400 mg in the TP was due to the solubility of the active substance, whereas the formulation
effect was based on an indirect observation that this was not the case for the RP.
Moreover, BE results should prevail over dissolution data and the 200 mg strength of the TP was BE to
the 200 mg strength of the RP, inasmuch as the 400 mg strength of the TP was BE to the 400 mg
strength of the RP.
Finally, a bracketing approach could be applicable in this situation since studies were available at the
extreme of the strength interval (200 and 400 mg).
Overall in vivo and in vitro evidence provided points to a positive answer to this question: a multiple
dose study in the highest strength can be considered sufficient to demonstrate bioequivalence despite
differences in the dissolution profiles (which can be explained because the dissolution profiles become
similar when tested at the same dose level per vessel), in case where a single-dose study can be
waived because of safety reasons, taking also into consideration the demonstrated BE in the single
dose study with the 200 mg strength and a bracketing approach between the 200 and 400 mg
strengths. This conclusion cannot be generalised and a case by case approach will be needed in similar
situations.
2. I s it acceptable and/ or needed to include early time points of the dissolution profiles in
the calculation of f2 values for a prolonged release tablet? Because f2 values are sensitive to
the choice of dissolution time points, what recommendations can be made for prolonged
release tablets in order to reliably conclude that the dissolution profiles can be considered
similar?
PKWP response

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 43/47

The design of a study comparing two dissolution profiles should take into account, among other
factors, the inclusion of relevant sampling time points. It is perfectly reasonable to use 2 h as a first
time point in a dissolution test running over 24 h. In the case at hand at 2 h already a relevant amount
(10 to 15 %) of the active has been released. On the other hand, early time points, even in the case of
a sustained release dosage form, are important in revealing release differences between the products
under comparison, because the mechanism controlling the release of the active substance is present
from the start.
Moreover, even though the choice of sampling time points could be questioned, there is no scientific
reason to exclude valid data in a calculation.
Therefore the PKWP was of the opinion that in this case the 2 h time point should not be omitted not
only because there was no scientific reason to exclude it but because the amount released was
considered relevant.
The choice of early time points in a comparative dissolution profile test should be based on the
relevance (mainly amount released and release controlling mechanism). On the other hand, the
conditions stated in Appendix 1 of the BE GL (2) should be complied with, namely
A minimum of three time points (zero excluded)
The time points should be the same for the two formulations
Twelve individual values for every time point for each formulation
Not more than one mean value of > 85% dissolved for any of the formulations.
The relative standard deviation or coefficient of variation of any product should be less than
20% for the first point and less than 10% from second to last time point.

________________________________________
(1) Note for guidance on modified release oral and transdermal dosage forms: Section II (pharmacokinetic and
clinical evaluation) CPMP/EWP/280/96
(2) Guideline on the investigation of bioequivalence CPMP/EWP/QWP/1401/98 Rev. 1/ Corr **

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 44/47

18. BCS classification of memantine
The PKWP was asked whether the drug substance memantine hydrochloride can be classified as BSC-
class I compound and therefore the framework of a BCS-based biowaiver in accordance with Appendix
III of the Guideline on the Investigation of Bioequivalence (CPMP/EWP/QWP/1401/98 Rev. 1) can be
applied.
Based on data on solubility and absorption/permeability characteristics of the drug substance, the
PKWP considers that the classification of memantine hydrochloride as BCS-class I compound (highly
soluble and highly permeable) is justified. The framework of a BCS-based biowaiver, which is in
general applicable to immediate release, solid pharmaceutical products for oral administration and
systemic action having the same pharmaceutical form, can therefore be used to address the question
of bioequivalence between specific test and reference products. The additional requirements specific for
BCS-class I compounds with regard to the drug product (in vitro dissolution and excipients) are
applicable in accordance with the above-mentioned guideline.

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 45/47

19. Ebastine: use of metabolite data to demonstrate bioequivalence
between inactive pro-drugs NEW
Background
The Guideline on the Investigation of Bioequivalence (CPMP/EWP/QWP/1401/98 Rev.1), states that:
Inactive pro-drugs
Also for inactive pro-drugs, demonstration of bioequivalence for parent compound is recommended.
The active metabolite does not need to be measured. However, some pro-drugs may have low plasma
concentrations and be quickly eliminated resulting in difficulties in demonstrating bioequivalence for
parent compound. In this situation it is acceptable to demonstrate bioequivalence for the main active
metabolite without measurement of parent compound. In the context of this guideline, a parent
compound can be considered to be an inactive pro-drug if it has no or very low contribution to clinical
efficacy.
In view of the above, and regarding the use of metabolite data to demonstrate bioequivalence between
inactive pro-drugs, the CMDh sought the Pharmacokinetics Working Partys (PKWP) opinion on the
following question:
Is it acceptable for a generic application for ebastine to demonstrate bioequivalence based
on either the parent ebastine or on the active metabolite carebastine, provided proper
justification in the study protocol has been provided, or can only one of these analytes be
used?
PKWP response
In the context of the Guideline on the investigation of bioequivalence (Doc. Ref.:
CPMP/QWP/EWP/1401/98 Rev. 1), the parent compound ebastine can be considered to be an inactive
pro-drug as it has no or very low contribution to clinical efficacy
1-6
.
Although demonstration of bioequivalence for parent compound is recommended for inactive pro-
drugs, demonstration of bioequivalence with ebastine would only be possible by inclusion of a very
high number of subjects. Indeed, ebastine has very low plasma concentrations, is rapidly and
extensively metabolised resulting in highly variable plasma concentrations of the parent compound,
resulting in a higher variability in pharmacokinetics than carebastine.
Therefore, bioequivalence studies using carebastine for bioequivalence evaluation would be considered
acceptable to detect formulation related differences between a test and a reference.
In summary, in accordance with the Guideline on the investigation of bioequivalence, it would be
acceptable to demonstrate bioequivalence based on the pharmacokinetics of the active metabolite
carebastine. However, in case an application is submitted solely with data on the parent ebastine, it is
also acceptable to demonstrate bioequivalence based on the pharmacokinetics of the parent ebastine.
In case both ebastine and carebastine are analysed, the analyte to be used for bioequivalence
evaluation should be prospectively defined in the protocol.
1. T. Yamaguchi, T. Hashizume, M. Matsuda, M. Sakashita, T. Fujii, Y. Sekine, M. Nakashima, and
T. Uematsu. Pharmacokinetics of the H1-receptor antagonist ebastine and its active metabolite
carebastine in healthy subjects. Arzneim.-Forsch. (1994) 44(1):59-64
2. Martinez-Tobed A., Tarrs E., Segura J., Roberts D.J. Pharmacokinetic studies of ebastine in
rats, dogs and man. Drugs Today (1992) 28(Suppl. B):57-67

Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 46/47

3. Wiseman L.R., Faulds D. Ebastine: a review of its pharmacological properties and clinical
efficacy in the treatment of allergic disorders. Drugs (1996) 51(2):260-277
4. Presa I.J. H1-antihistamines: a review. Alergol. Immunol. Clin. (1999) 14(5):300-312
5. Rico S., Antonijoan R.M., Barbanoj M.J. Ebastine in the light of CONGA recommendations for
the development of third-generation antihistamines. J. Asthma Allergy (2009) 2:73-92
6. Wood-Baker R., Holgate S.T. Dose-response relationship of the H1-histamine antagonist,
ebastine, against histamine and metacholine-induced bronchoconstriction in patients with asthma.
Agents and Actions (1990) 30:1-2


Questions & Answers: Positions on specific questions addressed to the
Pharmacokinetics Working Party

EMA/618604/2008 Rev. 9 Page 47/47

Das könnte Ihnen auch gefallen